Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Viral Immunol ; 36(6): 401-408, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37504965

RESUMEN

In this study, we reported the expression and potency of the recombinant H1N1 hemagglutinin (HA) vaccine as our in-house vaccine in a BALB/c mouse model. Recombinant H1N1 HA was produced in SF9 cell line, purified and formulated in MF59 adjuvant. Experimental mice were injected on days 0 and 14 with MF59-formulated vaccine, alum-based vaccine, and phosphate-buffered saline (PBS). Interleukin (IL)-2, IL-4, and interferon (IFN)-γ were assessed with commercial enzyme-linked immunosorbent assay (ELISA). Antibody responses and cytotoxic T lymphocyte (CTL) activity were assessed by hemagglutination inhibition and granzyme B ELISA, respectively. Moreover, the mice were challenged to show the vaccine efficacy. A considerable rise in IFN-γ and IL-4, as well as IFN-γ/IL-4 ratio, was observed in comparison with the alum-based vaccine and PBS group. Furthermore, our candidate vaccine showed superiority in humoral immune responses and CTL activity versus the alum-based vaccine and PBS group. The challenge showed that the survival rate in the vaccinated groups revealed a significant increase as compared with that in the PBS group. In conclusion, our candidate vaccine showed a robust Th1 response and CTL activity the alum-based vaccine. Moreover, a significant humoral immune response and a higher survival rate were detected in our vaccine as compared with the alum-based vaccine. It seems that the superiority of the MF59-based vaccine is due to the type of vaccine formulation in the candidate vaccine.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Interleucina-4 , Animales , Ratones , Citocinas , Hemaglutininas , Ratones Endogámicos BALB C , Adyuvantes Inmunológicos/farmacología , Vacunas Sintéticas
2.
Viral Immunol ; 36(6): 409-423, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37506342

RESUMEN

Despite the great success of vaccines in various infectious diseases, most current vaccines are not effective enough, and on the contrary, clinically approved alum adjuvants cannot induce sufficient immune responses, including a potent cellular immune response to confer protection. In this study, we used Nanochelating Technology to develop novel nanoadjuvants to boost the potency of the alum-adjuvanted inactivated severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccine. BALB/c mice were immunized twice over 2 weeks with different doses of adjuvanted-vaccine formulations and immune responses were assessed. The analysis results of IFN-γ and IL-17 cytokines demonstrated the effectiveness of the nanoadjuvants produced by the Nanochelating Technology in shifting the alum-based vaccine toward a stronger Th1 pattern. In addition, these nanoadjuvants improved IL-2 cytokine response, which shows the efficacy of these novel formulations in inducing specific T lymphocyte proliferation. Using these nanoadjuvants increased IL-10 cytokine secretion that may be representative of a better immunoregulatory impact and may also potentially prevent immunopathology responses. Moreover, specific IgG titer analysis revealed the potency of these nanoadjuvants in improving humoral immune responses. The enzyme-linked immunosorbent assay of receptor-binding domain (RBD)-specific IgG response showed that the developed novel formulations induced strong IgG responses against this protein. This study shows that the nanostructures produced by the Advanced Nanochelating Technology have potent adjuvant effects on alum-based SARS-CoV-2 vaccines to not only compensate for alum weakness in inducing the cellular immune responses by smart regulation of the immune system but also significantly improve the humoral and cellular immune responses simultaneously.


Asunto(s)
COVID-19 , Citocinas , Animales , Ratones , Humanos , SARS-CoV-2 , Vacunas de Productos Inactivados , Vacunas contra la COVID-19 , COVID-19/prevención & control , Adyuvantes Inmunológicos , Inmunoglobulina G , Anticuerpos Antivirales , Ratones Endogámicos BALB C
3.
Microb Pathog ; 176: 106007, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36709850

RESUMEN

Staphylococcus aureus is a gram-positive bacterium, representing one of the most important nosocomial pathogens. The treatment of infections, caused by S. aureus, has become increasingly intricate due to the emergence of highly resistant strains. Therefore, it is obvious that an effective prevention strategy against this bacterium could significantly decrease such infections. In the present study, the protective efficacy and immunological properties of recombinant autolysin, formulated in Montanide ISA266 and Alum adjuvants with Glucomannan as a polysaccharide, were assessed in the systemic mouse model of infection. Mice were immunized with the purified recombinant protein in various formulations in different groups and, subsequently, mice were challenged with 5 × 108 CFU of bacteria for the evaluation of their survival and bacterial clearances in the internal organs. ELISA was performed to determine the type of induced immunity, cytokine secretion (IFN-γ, IL-4, IL-2, and IL-17), and isotyping (IgG1 and IgG2a). In addition, we measured the opsonophagocytic activities of the antibodies. Results showed that immunization with r-autolysin + Alum + Glucomannan and r-autolysin + MontanideISA266+Glucomannan formulations significantly increased total IgG and isotypes (IgG1 and IgG2a), as compared with other vaccinated and control groups. Furthermore, the formulation of r-autolysin in Alum and MontanideISA266 adjuvants with Glucomannan enhanced IFN-γ, IL-4, and IL-17 cytokine secretion as well as protectivity, following experimental challenge. We concluded that Glucomannan has the potential to induce immune responses and would be used as an adjuvant factor in vaccine formulation.


Asunto(s)
Interleucina-17 , Staphylococcus aureus , Animales , Ratones , Interleucina-4 , N-Acetil Muramoil-L-Alanina Amidasa , Adyuvantes Inmunológicos , Adyuvantes Farmacéuticos , Mananos , Proteínas Recombinantes , Inmunoglobulina G , Inmunidad , Ratones Endogámicos BALB C
4.
IEEE Trans Nanobioscience ; 22(2): 393-400, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35914048

RESUMEN

Klebsiella pneumoniae can cause destructive changes to human lungs if aspirated. The present study aimed to evaluate the immunogenicity of the carriers of Poly lactic-co-glycolic acid (PLGA) and Methoxypoly(ethylene glycol) Poly(caprolactone) (MPEG-PCL) nanoparticles containing the capsular antigen of Klebsiella pneumoniae K2O1 in a model of pulmonary infection in mice as a vaccine candidate. Capsule antigen was extracted from K.pneumoniae K2O1 strain 1053 ATCC 10031 and transported with PLGA or MPEG-PCL nanoparticles as a vaccine in an animal model. The results of FT-IR and AFM confirmed the presence of antigen functional groups in the nanoparticle structure, and semi-spherical shape of the nanoparticles, respectively. The capsular polysaccharide was also used to evaluate the febrileness of the designed vaccine candidates based on the rabbits' pattern, and mortality due to the vaccine candidates in the mice. No fever was observed, and no mortality was observed in the mice. According to the results, the vaccine candidates designed to control the cause of pulmonary infections were effective in the liver, spleen, and lungs of the animals with the ability to enter the first stage of the clinical trial phase.


Asunto(s)
Klebsiella pneumoniae , Nanopartículas , Ratones , Humanos , Animales , Conejos , Espectroscopía Infrarroja por Transformada de Fourier , Portadores de Fármacos/química , Poliésteres/química , Polietilenglicoles/química , Nanopartículas/química
5.
Iran J Basic Med Sci ; 25(11): 1326-1333, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36474577

RESUMEN

Objectives: Here, immune responses and long-lived IgG responses of HBsAg-Alum, HBsAg-MF59, as well as HBsAg-MF59 were compared when formulated with PPD. Materials and Methods: BALB/c mice were vaccinated subcutaneously three times with a two-week -interval. Then, specific IgG, long-lived IgG responses up to 220 days, and IgG1/IgG2a isotypes, and IFN-γ and IL-4 on spleen cell culture supernatant were assessed using ELISA. Results: IFN-γ cytokine response between MF59- and Alum-adjuvanted vaccines did not show a significant difference. HBsAg-Alum revealed an increase in IL-4 cytokine versus HBsAg-MF59 at borderline (P=0.0553). In addition, HBsAg-MF59+PPD 10 µg showed a significant decrease in IL-4 and IFN-γ cytokines versus HBsAg-MF59. Furthermore, HBsAg-MF59+PPD10 µg showed a significant increase in the IL-2/IL-4 ratio versus HBsAg-MF59 (P=0.0339). Specific IgG antibody showed a significant increase in HBsAg-MF59, as compared with HBsAg-Alum. Furthermore, HBsAg-MF59 plus PPD showed a significant increase in IgG responses versus HBsAg-MF59 and HBsAg-Alum groups. Long-lived IgG responses showed a significant increase in HBsAgMF59 versus HBsAg-Alum group and PPD in the HBsAg-MF59 vaccine formulation, resulting in a significant increase in IgG responses versus HBsAg-MF59 group. In addition, HBsAg-MF59 plus PPD suppressed IgG1 response versus HBsAg-Alum. However, HBsAg-MF59 showed a significant increase in IgG2α versus the HBsAg-Alum group (P=0.0190). Immunization with HBsAg-MF59+PPD (10 µg) showed a significant increase versus the HBsAg-MF59 group (P=0.0040). IgG2a/IgG1 ratio in HBsAg-MF59+PPD1µg and HBsAg-MF59+PPD10 µg groups showed a significant increase versus HBsAg-MF59 groups (P<0.0345). Conclusion: PPD leads to a more potent long-lived IgG responses in the HBsAg vaccine, highlighting its potential as a component of a complex adjuvant.

6.
Viral Immunol ; 35(10): 663-672, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36534465

RESUMEN

Several inactivated SARS-CoV-2 vaccines have been approved for human use, but are not highly potent. In this study, different formulations of the inactivated SARS-CoV-2 virus were developed in Alum, Montanide 51VG, and Montanide ISA720VG adjuvants, followed by assessment of immune responses. The SARS-CoV-2 virus was inactivated with formalin and formulated in the adjuvants. BALB/c mice were immunized subcutaneously with 4 µg of vaccines on days 0 and 14; (IL-4) and (IFN-g), cytotoxic T lymphocyte (CTL) activity, and specific immunoglobulin G (IgG) titer and IgG1, IgG2a, and IgG2a/IgG1 ratio, and anti-receptor-binding domain (RBD) IgG response were assessed 2 weeks after the final immunization. Immunization with SARS-CoV-2-Montanide ISA51VG showed a significant increase in the IFN-γ cytokine versus SARS-CoV-2-Alum, SARS-CoV-2-Montanide ISA720VG, and control groups (p < 0.0033). Cytokine IL-4 response in SARS-CoV-2-Alum group showed a significant increase compared with SARS-CoV-2-Montanide ISA51VG, SARS-CoV-2-Montanide ISA720VG, and control groups (p < 0.0206). In addition, SARS-CoV-2-Montanide ISA51VG vaccine induced the highest IFN-γ/IL-4 cytokine ratio versus other groups (p < 0.0004). CTL activity in SARS-CoV-2-Montanide ISA51VG and SARS-CoV-2-Montanide ISA720VG groups showed a significant increase compared with SARS-CoV-2-Alum and control groups (p < 0.0075). Specific IgG titer in SARS-CoV-2-Montanide ISA51 VG and SARS-CoV-2-Montanide ISA720VG showed a significant increase compared with SARS-CoV-2-Alum and control groups (p < 0.0143). Results from specific IgG1and IgG2a in SARS-CoV-2-Alum, SARS-CoV-2-Montanide ISA51VG, and SARS-CoV-2-Montanide ISA720VG vaccine showed a significant increase compared with phosphate buffer saline (PBS) group (p < 0.0001), but SARS-CoV-2-Montanide ISA51VG and SARS-CoV-2-Montanide ISA 720VG groups showed the highest IgG2a/IgG1 ratio and a significant increase compared with SARS-CoV-2-Alum group (p < 0.0379). Moreover, inactivated SARS-CoV-2+Alum and SARS-CoV-2-Montanide ISA 720VG groups demonstrated a significant increase in anti-RBD IgG response versus the SARS-CoV-2-Montanide ISA51VG group. It seems that the type of vaccine formulation is a critical parameter, influencing the immunologic pattern and vaccine potency and human-compatible oil-based adjuvants were more potent than Alum adjuvant in the vaccine formulation.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra la COVID-19 , COVID-19 , Animales , Humanos , Ratones , Adyuvantes Inmunológicos/farmacología , COVID-19/prevención & control , Vacunas contra la COVID-19/química , Citocinas , Inmunidad , Inmunoglobulina G , Interferón gamma , Interleucina-4 , Ratones Endogámicos BALB C , SARS-CoV-2
7.
Biomed Environ Sci ; 35(9): 792-803, 2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36189994

RESUMEN

Objective: This study aimed to investigate the effects of Montanide ISA-720 and Naloxone (NLX) in Hepatitis B surface antigen (HBsAg) vaccine formulation on cytokine and long-lasting antibody responses. Methods: First, the HBsAg was formulated in Montanide ISA-720 adjuvant and Naloxone at 5 and 10 mg/kg. The experimental mice were immunized three times at a 2-week interval, and then IL-4, IL-2, TNF-α, and IFN-γ cytokines; long-lasting IgG antibody responses 220 days after the last shot; and IgG1/IgG2a isotypes were assessed by ELISA. Results: The HBsAg-Alum group exhibited the highest IL-4 cytokine response among the experimental groups, whereas NLX in HBsAg-MON720 vaccine formulation did not affect cytokine responses. In addition, NLX in Alum-based vaccine suppressed IL-4 cytokine response and increased the IL-2/IL-4 cytokine ratio. Moreover, HBsAg-MON720 was more potent than HBsAg-Alum in the induction of antibody responses, and NLX in Alum- and MON720-based vaccines induced long-lasting antibody responses. Conclusion: NLX in Alum-based vaccine decreased IL-4 cytokine response, increased IL-2/IL-4 cytokine ratio, and improved long-lasting humoral immune responses in both vaccine formulations. Therefore, the adjuvant activity of NLX in the vaccine formulation depends on the type of adjuvant and the nature of the antigen in the vaccine formulation.


Asunto(s)
Antígenos de Superficie de la Hepatitis B , Inmunidad Humoral , Adyuvantes Inmunológicos/farmacología , Compuestos de Alumbre , Animales , Citocinas , Vacunas contra Hepatitis B , Inmunoglobulina G , Interleucina-2 , Interleucina-4 , Ratones , Ratones Endogámicos BALB C , Aceite Mineral , Naloxona/farmacología , Factor de Necrosis Tumoral alfa
8.
Vaccines (Basel) ; 10(10)2022 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-36298611

RESUMEN

Konjac glucomannan (KGM) is a water-soluble polysaccharide derived from the Amorphophallus's tuber and, as herbal medicine has shown, can suppress tumor growth or improve health. However, there has been no investigation into the effects of KGM on breast tumor-bearing mice. Therefore, in two cohort experiments, we assessed the effect of glucomannan at daily doses of 2 and 4 mg for 28 days as a dietary supplement and also glucomannan in combination with tumor lysate vaccine as an adjuvant. Tumor volume was monitored twice weekly. In addition, TNF-α cytokines and granzyme B (Gr-B) release were measured with ELISA kits, and IL-2, IL-4, IL-17, and IFN-γ were used as an index for cytotoxic T lymphocyte activity. Moreover, TGF-ß and Foxp3 gene expression were assessed in a real-time PCR test. The results show that glucomannan as a dietary supplement increased the IFN-γ cytokine and Th1 responses to suppress tumor growth. Glucomannan as a dietary supplement at the 4 mg dose increased the IL-4 cytokine response compared to control groups. In addition, cell lysate immunization with 2 or 4 mg of glucomannan suppressed tumor growth. As an adjuvant, glucomannan at both doses showed 41.53% and 52.10% tumor suppression compared with the PBS group. Furthermore, the administration of glucomannan as a dietary supplement or adjuvant reduced regulatory T cell response through decreasing TGF-ß and Foxp3 gene expression in the tumor microenvironment. In conclusion, glucomannan as a dietary supplement or adjuvant enhanced the immune responses of tumor-bearing mice and decreased immune response suppression in the tumor milieu, making it a potentially excellent therapeutic agent for lowering breast tumor growth.

9.
Exp Parasitol ; 243: 108404, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36265591

RESUMEN

Toxoplasma gondii (T. gondii) remains as one of the controversial infections in the world. T. gondii is an important obligate intracellular protozoan parasite in the immune-deficient patients and pregnant women, sometimes leading to death and abortion, respectively. Herein, the adjuvant activity of nanocurcumin was assessed in the T. gondii killed vaccine model in BALB/c mice. In this study, 144 BALB/c mice were included in 8 groups and administered with different regimens of the vaccine; vac+30, 20 mg/kg of curcumin and nanocurcumin, vac + Freund's adjuvant, killed vac, vac + Alum adjuvant, and PBS via the subcutaneous route of immunization for three times with two-week intervals. Two weeks after the last immunization, the splenocytes' culture supernatant was evaluated for IL-4, IFN-γ, IL-2 and TNF-α cytokines and IFN-γ/IL-4, IFN-γ/TNF-α, and IL-2/IL-4 cytokine ratios using commercial ELISA kits. Specific total IgG antibodies, IgG1, and IgG2a were assessed with an optimized ELISA. Then the survival rate was determined 10 days after the experimental challenge. The results showed that the vaccine formulation in nanocurcumin at 20 mg/kg significantly increases IFN-γ cytokine and IFN-γ/IL4, IFN-γ/TNFα, and IL-2/IL4 ratios versus the vaccine formulated in curcumin, killed vaccine, and PBS group. In addition, specific total IgG antibody response showed that the vaccine formulated in nanocurcumin was more potent than that formulated in curcumin in the induction of humoral immune responses. Furthermore, results from the experimental challenge showed that nanocurcumin at a dose of 20 mg/kg could promote the life span of mice approximately by 12% versus the killed vaccine group. The present study showed that nanocurcumin in the vaccine formulation not only is more bioactive than curcumin in the modulation of cellular and humoral immune responses, but also provides more protectivity rate in the vaccinated mice on the killed T. gondii vaccine model. It seems that nanocurcumin can be used as an immunomodulator in vaccine formulation or as part of a complex adjuvant.


Asunto(s)
Adyuvantes Inmunológicos , Curcumina , Vacunas Antiprotozoos , Toxoplasma , Animales , Ratones , Anticuerpos Antiprotozoarios , Antígenos de Protozoos , Curcumina/farmacología , Citocinas , Inmunoglobulina G , Interleucina-2 , Interleucina-4 , Ratones Endogámicos BALB C , Proteínas Protozoarias , Factor de Necrosis Tumoral alfa , Vacunas de Productos Inactivados
10.
Iran J Basic Med Sci ; 25(5): 554-561, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35911642

RESUMEN

Objectives: SARS-CoV-2, emerging as a major threat to public health, has to be controlled through vaccination. Naloxone (NLX), an opioid receptor antagonist, demonstrated its adjuvant activity for microbial vaccines. In this study, inactivated SARS-CoV-2 was developed in the Alum/NLX adjuvant to increase the potency of the inactivated SARS-CoV-2 vaccine. Materials and Methods: BALB/c mice were immunized on days 0 and 14 with inactivated SARS-CoV-2-Alum, -Alum + NLX 3 mg/kg, -Alum + NLX 10 mg/kg, and -Freund adjuvant, as well as PBS. IFN-γ and IL-4 cytokines and Granzyme-B release were assessed with ELISA. In addition, specific total IgG, IgG1/IgG2a isotypes, and ratio as well as anti-RBD IgG responses were assessed with an optimized ELISA. Results: SARS-CoV-2-Alum-NLX10 group showed a significant increase in the IFN-γ cytokine response versus SARS-CoV-2-Alum, SARS-CoV-2-Alum-NLX3, and PBS groups. The SARS-CoV-2-Alum-NLX3 group exhibited a significant decrease in IL-4 cytokine versus SARS-CoV-2-Alum. The mice immunized with SARS-CoV-2-Alum-NLX10 showed a significant increase in CTL activity versus SARS-CoV-2-Alum and PBS. In addition, mice immunized with SARS-CoV-2-Alum-NLX3, SARS-CoV-2-Alum-NLX10 and SARS-CoV-2-Freund demonstrated an increase in IgG response, as compared with SARS-CoV-2-Alum and PBS group. Furthermore, all formulations of SARS-CoV-2 vaccines could induce both IgG1 and IgG2a isotypes. But, the IgG2a/IgG1 ratio in SARS-CoV-2-Freund and SARS-CoV-2-Alum-NLX10 revealed an increase as compared with that of the SARS-CoV-2-Alum group. Anti-RBD IgG response in the SARS-CoV-2-Alum-NLX10 group showed a significant increase as compared with the Alum-based vaccine. Conclusion: Formulation of inactivated SARS-CoV-2 virus in NLX/alum adjuvant improved the potency of humoral and, especially, cellular responses.

11.
Res Pharm Sci ; 17(4): 360-371, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36034086

RESUMEN

Background and purpose: Acinetobacter baumannii (A. baumannii) is known as a pathogen with antibiotic resistance, causing respiratory infections. PLGA has been approved for use in vaccines as well as drug delivery. This study was performed to evaluate PLGA nanoparticles containing the outer membrane proteins (OMPs) of A. baumannii in stimulating the mice's immune system and improving pneumonia. Experimental approach: Double emulsion solvent evaporation technique was used. The properties of the obtained nanospheres were determined using a zetasizer, FTIR, and AFM devices. Nanoparticles were administered to mice BALB/c by applying the intramuscular route. ELISA was used to measure the amounts of immunoglobulins produced; also, an opsonophagocytic killing assay was used to measure the effectiveness of immunoglobulins. Immunized mice were then challenged with live A. baumannii through the lungs; their internal organs were also removed for bacteriological studies. Findings/Results: The prepared particles were 550 nm in diameter with a negative surface charge. The production of the OMPs specific IgG was much higher in the group receiving nanoparticles containing antigen as compared to those getting pure antigen. The immunoglobulins produced against nanoparticles were superior to those developed against pure antigens. Mice that received the new nanovaccine were more resistant to pneumonia caused by this bacterium than those that received pure antigen. Conclusion and implication: Overall, it can be said that PLGA nanoparticles could deliver their internal antigens (OMPs) well to the immune system of mice and stimulate humoral immunity in these animals, thus protecting them against pneumonia caused by A. baumannii.

12.
Iran J Basic Med Sci ; 25(4): 442-450, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35656080

RESUMEN

Objectives: Methicillin-resistant Staphylococcus aureus (MRSA) reasons extreme infections, can resist various conventional antimicrobial agents, and cause morbidity and mortality worldwide. Vaccination seems to help modulate MRSA infections. Nanovaccine is considered a novel strategy in vaccine technology. The primary purpose of the present study was to develop a conjugate vaccine based on recombinant PBP2a and MRSA autolysin formulated in PLGA as a nanoparticle capable of enhancing protective responses against MRSA in the murine model. Materials and Methods: Recombinant PBP2a and autolysin have been expressed and purified by nickel-nitrilotriacetic acid (Ni-NTA) affinity column and characterized by SDS-PAGE and western blot. PLGA was bound to recombinant proteins by using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDAC) and adipic acid dihydrazide (ADH) as a linker and spacer, respectively. Conjugation of recombinant proteins to PLGA was confirmed by the AFM assay, zeta potential, and size distribution, and its efficacy was evaluated in mice. Total IgG, IgG1, IgG2a, IgG2b, and IgM titers were analyzed to assess immune responses. Lastly, the bioactivity of antibodies was tested by using the opsonophagocytosis assay. Results: Mice immunized with the r-PBP2a-r-autolysin-PLGA nanovaccine led to increased levels of opsonic antibodies and IgG1, IgG2a, IgG2b, and IgM when compared with other experimental groups. Our results confirmed that vaccination with nanovaccine could reduce the mortality rate against the sub-lethal dose of MRSA challenge. Furthermore, the nanovaccine could eliminate MRSA from the kidney of infected mice. Conclusion: This study may provide valuable insights into the protective power of the r-PBP2a-r-autolysin-PLGA conjugate vaccine against MRSA infection.

13.
Viral Immunol ; 35(3): 244-253, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35467429

RESUMEN

Declined immune response is the main cause of decreased potency of the influenza vaccine in the elderly, regardless of virus mutations. Herein, we hypothesized that the addition of α-tocopherol to the influenza vaccine formulation might increase vaccine potency and efficacy. Hemagglutinin of the H1N1 virus was formulated in Alum and α-tocopherol, and then aged (16-20-month-old) and young (6-8-week-old) mice were immunized subcutaneously two times with 2-week intervals with 5 µg of different vaccine formulations. Two weeks after the final boosting, IFN-γ and IL-4 cytokines were assessed by using ELISA. Humoral immune responses were assessed by hemagglutination inhibition (HI). In addition, vaccine efficacy was determined by intranasal viral challenge of mice using mouse-adapted H1N1 virus. Our results showed that the new vaccine formulation improved IFN-γ and IL-4 responses in the experimental mice. However, the increase was evident mainly in the aged group and, to some extent, in the young group. Results from the HI assay showed that α-tocopherol in the vaccine formulation could increase HI activity in both young and aged mice. Furthermore, α-tocopherol, as an adjuvant, increased the protectivity of the influenza vaccine in both aged and young groups through the decreased lung viral load and increased survival rate of the experimental mice. In conclusion, it seems that α-tocopherol can not only be used as an appropriate adjuvant for aged people, but also empower old and worn out cells to increase the effectiveness of the vaccine in the elderly.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Adyuvantes Inmunológicos , Anciano , Animales , Humanos , Inmunidad Humoral , Gripe Humana/prevención & control , Interleucina-4 , Ratones , Ratones Endogámicos BALB C , alfa-Tocoferol
14.
Viral Immunol ; 35(2): 150-158, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35319970

RESUMEN

New strategies to increase the immune response to HIV-1 vaccine using immunological adjuvants such as Toll-like receptor agonists are needed. In this study, HIV-1 p24-Nef and conjugated form of the vaccine candidate to type-A flagellin (FLA) were injected in the BALB/c mice in different routes. Two weeks after the last immunization, lymphocyte proliferation was measured by the BrdU method. The IL-4 and IFN-γ levels, as well as the total IgG antibody and its isotypes titer, were evaluated by the enzyme-linked immunosorbent assay method. The IFN-γ ELISPOT was also performed. Our data showed that the HIV-1 p24-Nef alone and conjugated to type-A flagellin (FLA) significantly increased lymphocyte proliferation responses as well as higher levels of cytokines and IFN-γ producing lymphocytes and the level of humoral immune responses compared with the control groups. The cell-mediated immune responses through the subcutaneous route and humoral immune responses through the intramuscular route were significantly higher in the conjugated form than in the mere vaccine candidate. In conclusion, when the FLA as an adjuvant is constructed in the HIV-1 vaccine candidate, it could effectively improve both humoral and cellular immune responses. Furthermore, modification in the vaccine formulation could change the optimal route of vaccine inoculation.


Asunto(s)
Vacunas contra el SIDA , VIH-1 , Adyuvantes Inmunológicos , Animales , Flagelina , Proteína p24 del Núcleo del VIH , Inmunización , Ratones , Ratones Endogámicos BALB C , Pseudomonas aeruginosa
15.
Vaccines (Basel) ; 10(2)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35214658

RESUMEN

Staphylococcus aureus (S. aureus) is an opportunistic pathogen that causes various inflammatory local infections, from those of the skin to postinfectious glomerulonephritis. These infections could result in serious threats, putting the life of the patient in danger. Antibiotic-resistant S. aureus could lead to dramatic increases in human mortality. Antibiotic resistance would explicate the failure of current antibiotic therapies. So, it is obvious that an effective vaccine against S. aureus infections would significantly reduce costs related to care in hospitals. Bacterial vaccines have important impacts on morbidity and mortality caused by several common pathogens, however, a prophylactic vaccine against staphylococci has not yet been produced. During the last decades, the efforts to develop an S. aureus vaccine have faced two major failures in clinical trials. New strategies for vaccine development against S. aureus has supported the use of multiple antigens, the inclusion of adjuvants, and the focus on various virulence mechanisms. We aimed to present a compressive review of different antigens of S. aureus and also to introduce vaccine candidates undergoing clinical trials, from which can help us to choose a suitable and effective candidate for vaccine development against S. aureus.

16.
Microb Pathog ; 160: 105184, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34508828

RESUMEN

Hepatitis B virus (HBV) infection is limited through vaccination against HBsAg formulated in the Alum adjuvant. However, this alum-formulated vaccine fails to be preventive in some cases, also known as non-responders. Recent studies have shown the immunomodulatory effect of α-tocopherol in various models. Here, we developed a new formulation for HBsAg using α-tocopherol, followed by assessment of immune responses. Experimental BALB/c mice were immunized with a commercial alum-based vaccine or the one formulated in α-tocopherol at different doses. Mice were immunized subcutaneously with 5 µg of HBsAg with different formulations three times with 2-week intervals. Specific total IgG, IgG1, and IgG2a isotypes of antibodies were measured by ELISA. Immunologic cytokines, such as IFN-γ, IL-4, IL-2, and TNF-α, were also evaluated through commercial ELISA kits. Our results showed that the new α-tocopherol-formulated vaccine had the ability to reinforce specific total IgG responses. Moreover, α-tocopherol in the HBsAg vaccine increased IFN-γ, IL-2, and TNF-α cytokines at higher concentrations; however, the vaccine suppressed IL-4 cytokine release. At a lower concentration of α-tocopherol, the IL-4 cytokine response increased without a positive effect on IFN-γ and TNF-α cytokine response. It seems that α-tocopherol can change the immune responses against HBsAg; however, the type of response depends on the dose of α-tocopherol used in the vaccine formulation.


Asunto(s)
Citocinas , Vacunas contra Hepatitis B , Interferón gamma/inmunología , Adyuvantes Inmunológicos , Animales , Citocinas/inmunología , Anticuerpos contra la Hepatitis B , Vacunas contra Hepatitis B/inmunología , Ratones , Ratones Endogámicos BALB C
17.
Iran J Basic Med Sci ; 24(4): 545-550, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34094038

RESUMEN

OBJECTIVES: Though immunization with HBsAg has been routine since the 1980s, it has numerous limitations such as low or none humoral immune responses. Today, nanotechnology is used in vaccinology to achieve higher potency. The present study deals with the achievement of fast antibody response of humoral immune responses using immune-targeting through mannosylated nanocarriers of the vaccine. MATERIALS AND METHODS: Mannose sugar and HBsAg were attached to the surface of iron oxide nanoparticles. Mannosylated iron oxide nanoparticles conjugated HBsAg (HBsAg +MLCMNP), iron oxide nanoparticles conjugated HBsAg (HBsAg +LCMNP), hepatitis B vaccine, and mere HBsAg were injected twice to BALB/c mice subcutaneously, while suitable control groups were considered. Specific total IgG antibodies were evaluated on the 7th and 14th days after the final immunization. The avidity maturation of the humoral immune response was assessed with an optimized ELISA. Graph pad prism software was used to analyze statistical data. RESULTS: Results showed that on the seventh day of the final shooting, the mannosylated nano-vaccine caused higher antibody response induction than nano-vaccine without mannose and commercial vaccine groups. After 14 days of the second injection, a significant difference was seen versus the nano-vaccine without mannose but not the commercial vaccine group. In addition, the avidity index in mannosylated nano-vaccine showed a significant increase compared with the nano-vaccine without mannose and mere HBsAg group but not compared with the commercial vaccine. CONCLUSION: It seems that mannosylated nano-vaccine has more potency to achieve fast antibody responses and also higher quality of humoral immune response.

18.
Microb Pathog ; 157: 104953, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34044042

RESUMEN

Methicillin resistant Staphylococcus aureus is one of the most common causes of nosocomial infections. Current therapeutic approaches are not always effective in treatment of nosocomial infections, thus, there is a global demand for the development of novel therapeutic strategies. Staphylococcus aureus possesses various systems to uptake iron. One of the most important of them is iron regulated surface determinant (Isd) which can be an excellent candidate for immunization. Here, following the preparation of recombinant IsdE protein, 20 µg of r-IsdE prepared in various formulations were subcutaneously injected in different groups of mice. Two booster vaccinations were administered in two-week intervals, then, blood samples were collected two weeks after each injection. ELISA was used for the evaluation of total IgG and its isotypes (IgG1 and IgG2a) as well as quantity of IFN-γ, IL-4, IL-17, IL-2 and TNF-α cytokines on the serum samples. Meanwhile, the immunized mice were intraperitoneally inoculated with 5 × 108 CFU of bacteria then, their mortality rate and bacterial load were assessed. Our results showed that immunization with the r-IsdE in various formulations raised total IgG and isotypes (IgG1 and IgG2a) compared with the control groups. Moreover, r-IsdE formulation with MF59 and Freund adjuvants raised production of IFN-γ, IL-4, IL-17, IL-2 and TNF-α cytokines and provided an acceptable protection against Staphylococcus aureus infections. Results of present study suggest that r-IsdE which can easily be expressed by Escherichia coli BL21 system shows a great potential to develop a protective immunity against infections caused by Methicillin resistant Staphylococcus aureus.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Vacunas , Animales , Clonación Molecular , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , Ratones Endogámicos BALB C , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/genética
19.
Probiotics Antimicrob Proteins ; 13(6): 1539-1545, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33843030

RESUMEN

Regulation of immune responses is among the beneficial effects of probiotic bacteria on human health. In this study, we aim to investigate the effect of normal and heat-shocked Lactobacillus plantarum PTCC 1058 cell lysate on cytokine expression by human PBMCs. The mid-exponential phase L. plantarum (108 CFU/mL) were used to prepare cell lysate. Isolated PBMCs were stimulated with 100 µg/mL of each normal and heat-shocked L. plantarum cell lysate for 72 h. Non-stimulated PBMCs were also evaluated as negative control. The mRNA expression of IL-6, IL-10, IFN-É£, TNF-α, and TGF-ß genes was determined by quantitative RT-PCR amplification of total RNA extracted from PBMCs. Both types of cell lysate were able to increase pro-inflammatory cytokines and decrease anti-inflammatory cytokines. However, this effect was significantly stronger in heat-shocked cell lysate-treated PBMCs. Moreover, comparison of IFN-É£/IL-10, IFN-É£/TGF-ß, IL-6/IL-10, IL-6/TGF-ß, and TNF-α/IL-10 ratios in both conditions demonstrated that in the heat-shocked group, all of the above ratios were significantly higher than normal lysate treatment (p˂0.001), suggesting that heat-shocked probiotics are a potent inducer of the immune system in comparison to intact probiotics. Regarding these results, it may be possible to develop a new postbiotic product for the stimulation of immune responses of cancer patients or individuals who suffer from an immune defect.


Asunto(s)
Citocinas/metabolismo , Lactobacillus plantarum , Leucocitos Mononucleares/inmunología , Células Cultivadas , Calor , Humanos , Interleucina-10 , Interleucina-6 , Factor de Crecimiento Transformador beta , Factor de Necrosis Tumoral alfa
20.
Int Immunopharmacol ; 96: 107599, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33848910

RESUMEN

Montanide ISA 51VG adjuvant has been approved for human clinical application and stimulates cellular and humoral immune responses. Here, HBsAg was formulated in Montanide ISA51VG adjuvant to compare its potency with the Fendrix and HBsAg-alum vaccines. In particular, the long-term humoral response was assessed up to 220 days after the final immunization. BALB/c mice were allocated into six groups. Treatment groups were injected with HBsAg-Montanide ISA51VG, the Fendrix and commercial HBsAg-alum, respectively. Montanide ISA51 VG, Alum and PBS injected mice were considered as control groups. Mice were immunized three times with 2-week intervals on days 0, 14 and 28 by subcutaneous injection. Lymphocyte proliferation was assessed with the BrdU method. IFN-γ, IL-2 and IL-4 cytokines, specific total IgG and IgG1/IgG2a isotypes were assessed using ELISA. The HBsAg-Montanide ISA51VG vaccine resulted in a significant increase in lymphocyte proliferation versus HBsAg-alum and higher IL-2 cytokine production versus the Fendrix. Comparable IL-4 and IFN-γ cytokines responses were observed for these vaccines. Following the first immunization, IgG increased more in HBs-Montanide 51VG group versus the HBs-alum group, while after the second and third shots comparable responses were observed in comparison to the HBs-alum group. Monitoring for 220 days after the final vaccination showed the superiority of HBsAg-Montanide ISA 51VG vaccine versus HBsAg-alum and even the Fendrix vaccine in the induction of long-term antibody responses. This study suggests that HBsAg-Montanide ISA51VG as a novel vaccine formulation can trigger both cellular and long-lasting humoral immune responses more efficiently than conventional HBsAg vaccines.


Asunto(s)
Composición de Medicamentos/métodos , Antígenos de Superficie de la Hepatitis B/inmunología , Inmunidad Humoral/inmunología , Manitol/análogos & derivados , Ácidos Oléicos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Formación de Anticuerpos/efectos de los fármacos , Citocinas/metabolismo , Femenino , Antígenos de Superficie de la Hepatitis B/administración & dosificación , Vacunas contra Hepatitis B/administración & dosificación , Vacunas contra Hepatitis B/inmunología , Inmunoglobulina G/sangre , Inyecciones Subcutáneas , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Manitol/administración & dosificación , Manitol/inmunología , Ratones Endogámicos BALB C , Ácidos Oléicos/administración & dosificación , Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...